Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Biomed Mater Res A ; 102(5): 1558-67, 2014 May.
Article in English | MEDLINE | ID: mdl-23775982

ABSTRACT

Treatment of nonunion fractures is a significant problem. Common therapeutics, including autologous bone grafts and bone morphogenetic proteins, show well-established limitations. Therefore, a need persists for the identification of novel clinical therapies to promote healing. The Notch signaling pathway regulates bone development. Clinically, loss-of-function mutations to the Notch ligand Jagged1 decrease bone mass and increase fracture risk. Jagged1 is also the most highly upregulated ligand during fracture repair, identifying it as a potential target to promote bone formation. Therefore, the objective of this study was to develop a clinically translatable construct comprised of Jagged1 and an osteoconductive scaffold, and characterize its activity in human mesenchymal stem cells (hMSC). We first evaluated the effects of Jagged1 directly immobilized to a novel poly(ß-amino ester) relative to indirect coupling via antibody. Direct was more effective at activating hMSC Notch target gene expression and osteogenic activity. We then found that directly immobilized Jagged1 constructs induced osteoblast differentiation. This is the first study to demonstrate that Jagged1 delivery transiently activates Notch signaling and increases osteogenesis. A positive correlation was found between Jagged1-induced Notch and osteogenic expression. Collectively, these results indicate that Jagged1 coupled to an osteogenic biomaterial could promote bone tissue formation during fracture healing.


Subject(s)
Calcium-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Osseointegration/drug effects , Osteogenesis/drug effects , Polymers/pharmacology , Receptors, Notch/metabolism , Signal Transduction/drug effects , Alkaline Phosphatase/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biocompatible Materials/pharmacology , Calcification, Physiologic/drug effects , Cell Count , Cell Cycle Proteins/metabolism , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Immobilized Proteins/metabolism , Jagged-1 Protein , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Phenotype , Rats , Serrate-Jagged Proteins , Tissue Scaffolds/chemistry
2.
PLoS One ; 8(7): e68726, 2013.
Article in English | MEDLINE | ID: mdl-23844237

ABSTRACT

The Notch signaling pathway is an important regulator of embryological bone development, and many aspects of development are recapitulated during bone repair. We have previously reported that Notch signaling components are upregulated during bone fracture healing. However, the significance of the Notch pathway in bone regeneration has not been described. Therefore, the objective of this study was to determine the importance of Notch signaling in regulating bone fracture healing by using a temporally controlled inducible transgenic mouse model (Mx1-Cre;dnMAML(f/-)) to impair RBPjκ-mediated canonical Notch signaling. The Mx1 promoter was synthetically activated resulting in temporally regulated systemic dnMAML expression just prior to creation of bilateral tibial fractures. This allowed for mice to undergo unaltered embryological and post-natal skeletal development. Results showed that systemic Notch inhibition prolonged expression of inflammatory cytokines and neutrophil cell inflammation, and reduced the proportion of cartilage formation within the callus at 10 days-post-fracture (dpf) Notch inhibition did not affect early bone formation at 10dpf, but significantly altered bone maturation and remodeling at 20dpf. Increased bone volume fraction in dnMAML fractures, which was due to a moderate decrease in callus size with no change in bone mass, coincided with increased trabecular thickness but decreased connectivity density, indicating that patterning of bone was altered. Notch inhibition decreased total osteogenic cell density, which was comprised of more osteocytes rather than osteoblasts. dnMAML also decreased osteoclast density, suggesting that osteoclast activity may also be important for altered fracture healing. It is likely that systemic Notch inhibition had both direct effects within cell types as well as indirect effects initiated by temporally upstream events in the fracture healing cascade. Surprisingly, Notch inhibition did not alter cell proliferation. In conclusion, our results demonstrate that the Notch signaling pathway is required for the proper temporal progression of events required for successful bone fracture healing.


Subject(s)
Bony Callus/metabolism , Fracture Healing/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Bone Remodeling/genetics , Bony Callus/pathology , Cartilage/metabolism , Cell Differentiation/physiology , Chondrogenesis/genetics , Female , Gene Expression , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics
3.
J Orthop Res ; 31(6): 935-43, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23280580

ABSTRACT

Thrombospondin-2 (TSP2) is a matricellular protein that is highly up-regulated during fracture healing. TSP2 negatively regulates vascularity, vascular reperfusion following ischemia, and cutaneous wound healing. As well, TSP2-null mice show increased endocortical bone formation due to an enhanced number of mesenchymal progenitor cells and show increased cortical thickness. Mice deficient in TSP2 (TSP2-null) show an alteration in fracture healing, that is unrelated to their cortical bone phenotype, which is characterized by enhanced vascularization with a shift towards an intramembranous healing phenotype; thus, we hypothesized that there would be enhanced ischemic fracture healing in the absence of TSP2. We investigated whether an absence of TSP2 would enhance ischemic fracture healing utilizing Laser doppler, µCT and histological analysis. Ischemic tibial fractures were created in wildtype (WT) and TSP2-null mice and harvested 10, 20, or 40 days post-fracture. TSP2-null mice show enhanced vascular perfusion following ischemic fracture. At day 10 post-fracture, TSP2-null mice have 115% greater bone volume than WT mice. This is associated with a 122% increase in vessel density, 20% increase in cell proliferation, and 15% decrease in apoptosis compared to WT. At day 20, TSP2-null mice have 34% more bone volume, 51% greater bone volume fraction, and 37% more bone tissue mineral density than WT. By 40 days after fracture the TSP2-null mice have a 24% increase in bone volume fraction, but other parameters show no significant differences. These findings indicate TSP2 is a negative regulator of ischemic fracture healing and that in the absence of TSP2 bone regeneration is enhanced.


Subject(s)
Extremities/blood supply , Fracture Healing , Neovascularization, Physiologic , Regional Blood Flow , Thrombospondins/physiology , Animals , Apoptosis , Bony Callus/blood supply , CD36 Antigens/metabolism , CD47 Antigen/metabolism , Cartilage/growth & development , Cell Proliferation , Ischemia/physiopathology , Mice , Mice, Inbred C57BL , Thrombospondin 1/metabolism
4.
J Orthop Res ; 30(2): 296-303, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21818769

ABSTRACT

Previous studies have demonstrated that Notch signaling regulates endochondral and intramembranous bone formation by controlling cell proliferation and differentiation. Notch signaling has also been shown to regulate healing in a variety of tissues. The objective of this study was to characterize and compare activation of the Notch signaling pathway during endochondral and intramembranous bone healing using tibial fracture and calvarial defect injury models, respectively. Bilateral tibial fractures or bilateral 1.5 mm diameter calvarial defects were created in mice, and tissues were harvested at 0, 5, 10, and 20 days post-fracture. Gene expression of Notch signaling components was upregulated during both tibial fracture and calvarial defect healing, with expression generally higher during tibial fracture healing. The most highly expressed ligand and receptor during healing, Jag1 and Notch2 (specifically the activated receptor, known as NICD2), were similarly localized in mesenchymal cells during both modes of healing, with expression decreasing during chondrogenesis, but remaining present in osteoblasts at all stages of maturity. Results suggest that in addition to embryological bone development, Notch signaling regulates both endochondral and intramembranous bone healing.


Subject(s)
Bone Regeneration , Receptors, Notch/physiology , Animals , Calcium-Binding Proteins/analysis , Fracture Healing , Intercellular Signaling Peptides and Proteins/analysis , Jagged-1 Protein , Male , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Receptor, Notch2/analysis , Serrate-Jagged Proteins , Skull/injuries , Tibial Fractures/physiopathology , Up-Regulation
5.
Stem Cells ; 28(6): 1071-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20506495

ABSTRACT

Leptin functions through a well-documented central neuroendocrine pathway to regulate bone mass. However, the ability of leptin to modulate bone mass through a peripheral mechanism has been debated due to conflicting in vitro results and lack of sufficient in vivo models. We utilized mice with LoxP sites introduced into the long-form leptin receptor (ObRb) gene to determine how leptin regulates mesenchymal progenitor cell (MPC) differentiation and osteoblast function in vitro and in vivo. Rapid phosphorylation of Stat3 after leptin treatment of bone marrow stromal cells (BMSCs) from mice with conditional deletion of ObRb in macrophages (LysM(Cre+F/F)) confirmed expression of functional leptin receptors by BMSCs. Adenovirus-Cre mediated disruption of ObRb in primary stromal cells decreased mineralization and increased adipogenesis. In contrast, BMSCs harvested from leptin-signaling deficient Ob/Ob or Db/Db mice showed increased mineralization. To determine the physiologic relevance of these differences, mice with cell-specific deletion of ObRb in mesenchymal precursors (3.6(Cre+F/F)) or osteoblasts (2.3(Cre+F/F)) were generated. Although the 2.3(Cre+F/F) mice were grossly normal, the 3.6(Cre+F/F) mice displayed mild obesity that was not attributed to food intake. Femurs of 3.6(Cre+F/F) animals showed a 58%-61.9% increase in trabecular bone volume and a 65.5%-74% increase in bone mineral density. Cortical volume and mineral content were also increased 18%-22%. Primary 3.6(Cre+F/F) BMSCs recapitulated the high mineralization phenotype of Ob/Ob and Db/Db BMSCs. We conclude that leptin may have multiple peripheral roles depending on the differentiation state of MPC. Leptin (a) helps maintain MPCs in an undifferentiated state and (b) promotes mineralization of more differentiated osteoblasts.


Subject(s)
Cell Differentiation , Leptin/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Receptors, Leptin/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Calcification, Physiologic , Cells, Cultured , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteoblasts/metabolism , Receptors, Leptin/genetics
6.
J Biomech ; 43(4): 778-82, 2010 Mar 03.
Article in English | MEDLINE | ID: mdl-19931082

ABSTRACT

Due to inadequate healing, surgical repairs of torn rotator cuff tendons often fail, limiting the recovery of upper extremity function. The rat is frequently used to study rotator cuff healing; however, there are few systems capable of quantifying forelimb function necessary to interpret the clinical significance of tissue level healing. We constructed a device to capture images, ground reaction forces and torques, as animals ambulated in a confined walkway, and used it to evaluate forelimb function in uninjured control and surgically injured/repaired animals. Ambulatory data were recorded before (D-1), and 3, 7, 14, 28 and 56 days after surgery. Speed as well as step width and length were determined by analyzing ventral images, and ground reaction forces were normalized to body weight. Speed averaged 22+/-6 cm/s and was not affected by repair or time. Step width and length of uninjured animals compared well to values measured with our previous system. Forelimbs were used primarily for braking (-1.6+/-1.5% vs +2.5+/-0.6%), bore less weight than hind limbs (49+/-5% vs 58+/-4%), and showed no differences between sides (49+/-5% vs 46+/-5%) for uninjured control animals. Step length and ground reaction forces of the repaired animals were significantly less than control initially (days 3, 7 and 14 post-surgery), but not by day 28. Our new device provided uninjured ambulatory data consistent with our previous system and available literature, and measured reductions in forelimb function consistent with the deficit expected by our surgical model.


Subject(s)
Disease Models, Animal , Forelimb/physiopathology , Gait , Rotator Cuff Injuries , Rotator Cuff/physiopathology , Tendon Injuries/physiopathology , Tendon Injuries/surgery , Animals , Foot/physiopathology , Humans , Male , Rats , Rats, Sprague-Dawley , Rotator Cuff/surgery , Stress, Mechanical , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...